Cerebrospinal fluid A beta 1-40 peptides increase in Alzheimer’s disease and are highly correlated with phospho-tau in control individuals ============================================================================================================================================ * Sylvain Lehmann * Julien Dumurgier * Xavier Ayrignac * Cecilia Marelli * Daniel Alcolea * Juan Fortea Ormaechea * Eric Thouvenot * Constance Delaby * Christophe Hirtz * Jérôme Vialaret * Nelly Ginestet * Elodie Bouaziz-Amar * Jean-Louis Laplanche * Pierre Labauge * Claire Paquet * Alberto Lleo * Audrey Gabelle * for the Alzheimer’s Disease Neuroimaging Initiative (ADNI) ## ABSTRACT **Background:** Amyloid pathology, which is one of the characteristics of Alzheimer’s disease (AD), results from altered metabolism of the beta-amyloid peptide (Aβ) in terms of synthesis, clearance or aggregation. A decrease in cerebrospinal fluid (CSF) level Aβ 1-42 is evident in AD, and the CSF ratio Aβ 40 /Aβ 40 has recently been identified as one of the most reliable diagnostic biomarkers of amyloid pathology. Variations in inter-individual levels of Aβ 1-40 in the CSF have been observed in the past, but their origins remain unclear. In addition, the variation of Aβ 40 in the context of AD studied in several studies has yielded conflicting results. **Methods** Here, we analyzed the levels of Aβ 1-40 using multicenter data obtained on 2466 samples from six different cohorts in which CSF was collected under standardized protocols, centrifugation and storage conditions. Tau and p-tau(181) concentrations were measured using commercially available in vitro diagnostic immunoassays. Concentrations of CSF Aβ 1-42 and Aβ 1-40 were measured by ELISA, xMAP technology, chemiluminescence immunoassay (CLIA) and mass spectrometry. Statistical analyses were calculated for parametric and non-parametric comparisons, linear regression, correlation and odds ratios. The statistical tests were adjusted for the effects of covariates (age, in particular). **Results:** Regardless of the analysis method used and the cohorts, a slight but significant age-independent increase in the levels of Aβ 40 in CSF was observed in AD. We also found a strong positive correlation between the levels of Aβ 40 and p-tau(181) in CSF, particularly in control patients. **Conclusions:** These results indicate that an increase in the baseline level of amyloid peptides, which are associated with an increase in p-tau(181), may be a biological characteristic of AD. This confirms the potential therapeutic value of lowering the baseline levels of Aβ 40 which, being elevated, can be considered a risk factor for the disease. Keywords * Alzheimer’s disease * amyloid peptides * tau proteins * Biomarkers * Cerebrospinal Fluid (CSF) * cerebrospinal fluid ## Background Alzheimer’s disease (AD) neuropathological brain lesions consist of aggregates of hyperphosphorylated tau proteins, which have also been called neurofibrillary tangles (NFTs), and extracellular deposits of amyloid precursor protein (APP) derived amyloid-beta peptides (Aβ), which are known as amyloid plaques. Much research has been focusing recently on the molecular mechanisms underlying these pathological events as it has become essential to develop preventive and therapeutic strategies for AD. For a long time, the main explanation for the pathogenesis of AD was that amyloidogenesis was the *primum movens* of the affection, which led to the concept of the amyloid cascade [1]. According to this picture of the disease, the alteration of APP metabolism (increasing amyloid production, decreasing clearance rates..), the aggregation of Aβ peptides and the formation of amyloid plaques, might result in microglial and astrocyte activation, local inflammatory responses, oxidative stress and eventually in the hyperphosphorylation of tau proteins and secondarily, in the formation of NFTs [2]. The idea that amyloid peptides contribute importantly to the etiology of AD is supported by cases of AD who carry presenilins (1 or 2) or APP mutations [3]. These gene mutations trigger the overproduction of Aβ peptides or the preferential production of Aβ42, which is the most amyloidogenic of all the peptides. An APP gene dose effect triggering AD development, as occurs in Down syndrome [4] and in gene duplication processes [5], is a further/an additional potential factor contributing to amyloid pathogenesis. Other genetic factors have been described, such as apolipoprotein E4 allele, in particular [6]. Studies on cerebrospinal fluid (CSF) biomarkers in AD have greatly improved our understanding of the pathophysiology of this disease. The production of amyloid peptides following the neuronal processing of APP has been involved in the response to physiological challenge with neurotrophic, anti-microbial, tumor suppression or synaptic function regulation activities [7]. Regarding tau proteins which are associated to microtubules, their physiological secretion by neuronal cells is a recent discovery which physiological relevance and benefit is still matter of debate [8]. A decrease in CSF Aβ 42 is especially indicative of an amyloidogenic process, while an increase in tau proteins (total tau and its phosphorylated form p-tau(181)) is known to be associated with axonal loss and tau pathology in AD [9, 10]. Tests on these two biomarkers are being included nowadays in the international clinical research guidelines [11, 12], and many centers [11–15], and ourselves [13–15] have integrated them into daily clinical practice. Importantly, these biochemical CSF measurements are concordant with the results of the PET imaging approaches which were initially developed to determine the brain amyloid load [16], and now also serve to measure tau accumulation [17]. These data are in line with hypotheses put forward by Jack *et al*, [18] about the chronology of the evolution of biomarkers during the pathophysiological process, and the relevance of amyloid markers in particular at a very early stage, probably as early as 10 to 15 years before the onset of clinical symptoms. Under non-pathological conditions, Aβ40 is highly correlated with Aβ42 [19]. The computation of the ratio Aβ42/40 is now being used in routine clinical practice on AD patients in some centers [20–22]. This is a useful approach for reducing pre-analytical Aβ42 biases [23–25] and improving the diagnostic performances of CSF biomarkers [26], especially in discordant cases [27]. This ratio can also be used to account for interindividual amyloid variations in the baseline CSF level [28]. Low CSF Aβ40 levels might also be indicative of frontotemporal dementia (FTD) [29, 30], cerebral amyloid angiopathy (CAA) [31], HIV [32], multiple sclerosis [33] or normal pressure hydrocephalus [34]. In their meta-analysis of AD biomarkers, Olsson et al. [35] observed the existence of a negligible difference in CSF Aβ40 between AD and control patients. Most of the 32 studies considered had a limited number of patients included in each group (the median number of subjects per group was less than 30, and the maximum number of subjects was 137 and 328 in AD and non-AD groups, respectively). The focus of these studies were also quite different, looking at the diagnostic interest of Aβ42/40 in AD, of of Aβ peptides in other neurodegenerative diseases, or being more interested in pathophysiological mechanisms. In the present study, we revisited the issue of the Aβ40 levels using large series of multicentre data. The results show the occurrence of a significant age-independent increase in CSF Aβ40 in AD. Another noteworthy finding was the existence of a strong positive correlation between CSF Aβ40 and the p-tau(181) concentration, even in patients without Alzheimer’s disease (NAD). These findings suggest that the baseline amyloid peptide level may constitute a risk factor contributing to sporadic AD, which is associated with p-tau(181) production. ## Methods ### Study design and subjects Patients with cognitive impairments were recruited and followed at the Montpellier and Paris Memory Resources Center (CMRR). The Montpellier participants were subdivided into two cohorts which were recruited during different periods: Montpellier 1 (Mtp-1) (recruited from 07/2015 to 05/2017) and Montpellier 2 (Mtp-2) (recruited from 09/2009 to 06/2015). These two periods corresponded to the use of different ELISA kits (SupTable 1). The cohort Mtp-1 consisted of 400 patients (126 AD, 274 NAD), the cohort Mtp-2 consisted of 504 patients (220 AD, 284 NAD). The Paris cohort consisted of 624 patients (299 AD, 325 NAD) from the Centre de Neurologie Cognitive, Groupe Hospitalier Lariboisière Fernand-Widal (recruited from 03/2012 to 05/2017). The Barcelona SPIN (Sant Pau Initiative on Neurodegeneration) cohort (79 AD, 148 NAD) consisted of patients who had undergone lumbar puncture for CSF AD biomarkers at the Sant Pau Memory Unit [36, 37] (recruited from 05/2009 to 12/2017). All the patients underwent a thorough clinical examination including biological lab tests, neuropsychological assessments and brain imaging. The same diagnostic procedure [27] and AD diagnostic criteria [38] were used at all the clinical centres which participated. The NAD diagnosis included FTD based on relevant criteria [39], dementia with Lewy bodies based on the McKeith criteria [40]), corticobasal degeneration (based on the criteria defined by Boeve et al [41]), progressive supranuclear palsy, amyotrophic lateral sclerosis, chronic hydrocephalus, vascular dementia and psychiatric disorders (based on the usual consensus diagnostic criteria). All the patients at each clinical centre gave their written informed consent to participating in clinical research on CSF biomarkers, which was approved by the respective Ethics Committees. The committee responsible in Montpellier was the regional Ethics Committee of the Montpellier University Hospital and Montpellier CSF-Neurobank #DC-2008-417 at the certified NFS 96-900 CHU resource center BB-0033-00031, [www.biobanques.eu](http://www.biobanques.eu). Authorization to handle personal data was granted by the French Data Protection Authority (CNIL) under the number 1709743 v0. Two sets of data originating from the analysis of CSF samples at the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database ([www.loni.ucla.edu/ADNI](http://www.loni.ucla.edu/ADNI)) were used after the agreement of the scientific committee. ADNI UPEN-RESULTS, UPEN-ELYCYS (n = 311) and MS UPENNMSMSABETA (n = 400) data were also used. In the ADNI cohorts, which included many patients with mild cognitive impairments (MCI), we had to rely on the biological PLM (Paris-Lille-Montpellier) scale [42] to define populations with a low (ADNI(-)) and high (ADNI(+)) prevalence of AD. This scale combines the concentration of the three CSF biomarkers [Aβ42, tau, ptau(181)] into a probability scale for AD. The score ranges from 0 to 3 based on the number of abnormal CSF biomarkers. ADNI(-) population corresponded to PLM scale of 0 or 1 with less than 25% of AD, while ADNI(+) corresponded to PLM scale of 2 and 3 with more than 75% of AD. Importantly the PLM score used was not based on the Aβ40 values so as to prevent circular reasoning. This way of stratifying patients in the ADNI cohort represents anyway a limitation of our study. ### CSF samples and assays CSF was collected using standard conditions of collection, centrifugation and storage [43, 44]. CSF tau and p-tau(181) concentrations were measured using the standardized commercially available INNOTESTR sandwich ELISA, Luminex® xMAP technology (x = analyte, MAP = Multi-Analyte Profiling) assays in line with the manufacturer’s instructions (Fujirebio-Europe). The consistency of the p-tau(181) detection using the ELISA assays is ensured by its comparison with the mass spectrometry detection performed in this fluid [45]. In the Mtp-1 cohort, CSF Aβ1-42 and Aβ1-40 (denoted here by Aβ42 and Aβ40) were measured with Euroimmun kits (EQ-6511-9601 (Aβ1-40); EQ-6521-9601 (Aβ1-42)). In the Mtp-2 and Paris cohorts, CSF Aβ42 and Aβ40 were measured using INNOTESTR sandwich ELISA from IBL and Fujirebio, respectively, as recommended by the manufacturer. Roche Elecsys automated chemiluminescence immunoassay (CLIA) and mass spectrometry were used on the ADNI cohorts to measure CSF Aβ1-42 and Aβ1-40 as previously described [46, 47]. Detection limits of these kits are compatible with CSF clinical ranges. Average concentration of analytes may differs between kits in relation with standard value assignments by the vendors in the absence of certified reference materials. The pre-analytical procedure was standardized [44] but differed, depending on the type of collection tubes used [36, 48]). This explains the differences observed between cohorts in the mean aβ40 and Aβ42 values measured with the same detection kit (SupTable 1). The quality of the results was ensured by using validated standard operating procedures and internal quality controls (QCs). The QC coefficient of variation obtained on the CSF analytes in each batch and between batches ranged consistently below 15%. In addition, external QC procedures were used to confirm the quality/accuracy of the results [43]. In the case of the ADNI cohorts, CSF samples were deep frozen after the lumbar puncture without performing any centrifugation or aliquoting, and shipped to the UPENN ADNI Biomarker Laboratory in Philadelphia on dry ice, where they were thawed, aliquoted, and re-frozen. ### Statistical analysis Statistical analyses were computed with the MedCalc software program (18.11.3). Data tested for normality are expressed as means ± SDs, and differences between groups were taken to be significant in the Student’s t-tests at P < 0.05. Linear regression was computed between continuous biomarkers, and the corresponding Pearson correlation coefficients and statistical significance have been specified in the tables. When indicated, statistical tests were adjusted to account for the effects of covariates (age, in particular). Odds ratios corresponded to the presence of AD in the various percentile groups, based on the distribution of Aβ40. The 95% confidence odds ratio intervals were computed along with the z-statistics and the associated P-values. ## Results ### CSF Aβ42 and Aβ40 in AD and NAD populations CSF data on 2466 samples originating from six different cohorts were included in the present study. The AD and NAD populations were defined based on clinical criteria in the Montpellier 1 (Mtp-1), Montpellier 2 (Mtp-2), Paris and Barcelona cohorts, and on the PLM score for the ADNI cohorts. Differences were observed in terms of age and CSF biomarker profiles in the overall population, as well as in each clinical cohort (Table 1). As was to be expected, AD patients obtained lower MMSE scores and showed higher CSF tau and p-tau(181) levels than NAD patients/participants. A significant decrease in the CSF Aβ42 concentrations was observed in the AD population (Figure 1A), regardless of the cohort tested. Noteworthy differences were also observed in the Aβ40 levels between all the cohorts (Figure 1B): the values recorded in the AD population were significantly higher than in the NAD group regardless of analytical method, the sex or the age as covariate. ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/02/2020.06.02.20119578/F1/graphic-1.medium.gif) [](http://medrxiv.org/content/early/2020/06/02/2020.06.02.20119578/F1/graphic-1) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/02/2020.06.02.20119578/F1/graphic-2.medium.gif) [](http://medrxiv.org/content/early/2020/06/02/2020.06.02.20119578/F1/graphic-2) Figure 1: CSF Aβ42 and Aβ40 in Non-AD and AD populations CSF concentration of Aβ42 (panel A) and Aβ40 (panel B) in six independent cohorts (Montpellier 1 (Mtp-1), Montpellier 2 (Mtp-2), Paris, SPIN-Barcelona, ADNI-MA, ADNIElecsys) confirmed the significant difference between NAD and AD patients for both analytes (ttest). Note that Aβ has been assess using five different detection methods (supTable 1). View this table: [Table 1](http://medrxiv.org/content/early/2020/06/02/2020.06.02.20119578/T1) Table 1 Demographical and cerebrospinal fluid (CSF) biomarkers characteristics of the six cohorts, Montpellier 1 (Mtp-1), Montpellier 2 (Mtp-2), Paris, SPIN-Barcelona, ADNI-MS and ADNIElecsys. Results are expressed as the mean +/− standard deviation (SD). Abbreviation: MMSE = mini mental state examination; AD = Alzheimer’s disease; NAD = Non Alzheimer’s disease; ADNI(-) = cognitive patients with non Alzheimer’s disease PLM profile; ADNI(+) = cognitive patients with Alzheimer’s disease PLM profile; P significance level of the Student’s t-test and * Chi-squared test for the comparison of two proportions. Values of Aβ40, Aβ42, tau and p-tau(181) are in pg/mL. In the cohorts that combined AD, MCI, FTD, Control (subjective cognitive impairment) and other neurological diseases (OND) patients, the difference between AD and the other clinical groups was eventually confirmed (Figure 2A). The influence of *APOE* status, which was available in the case of 983 samples (524 NAD with 36.6% E4+; 459 AD with 58.4% E4+), was also assessed with respect to the Aβ levels (SupFigure 1A-F). As previously reported [49, 50], the presence of ApoE4 was significantly associated with lower Aβ42 levels, as well as lower Aβ40 levels. The difference in Aβ40 values between NAD and AD patients was also observed in both ApoE4 positive and negative populations (SupFigure 1G-I). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/02/2020.06.02.20119578/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/06/02/2020.06.02.20119578/F2) Figure 2: Aβ40 in different diagnosis; Representation in percentile; AD odd ratio, age and p-tau(181) distribution The SPIN-Barcelona, Mtp-2 and Paris cohorts displayed a large range of pathological samples from patients with AD, Mild Cognitive Impairment (MCI), FTD, Control (subjective cognitive impairment) and other neurological diseases (OND). Mean-centred Aβ40 values in these cohorts were combined and compared in the different clinical groups confirming the significant increase of the peptides in AD (Panel A). The four cohorts (Montpellier 1 (Mtp-1), Montpellier 2 (Mtp-2), Paris, SPIN-Barcelona) have been sorted in four classes based on their Aβ40 percentile values as follows; p25: < 25th percentile, p25-50: 25th-50th percentile; p50-75: 50th-75th percentile; p75: > 75th. The odd ratio for AD (panel B), the age of the patients (panel C) and the concentration of CSF p-tau(181) (panel D) were then plotted in each percentile class. Significant differences between classes are indicated. To investigate more closely the relationship between Aβ40 and AD diagnosis, the total population was sorted into four percentile classes based on the value of this biomarker in the CSF (< 25th, 25th-50th, 50th-75th and > 75th percentiles). The percentage of AD patients in each cohort clearly increased along with the Aβ40 percentiles (SupTable2). To account for the differences in AD prevalence between the cohorts, the odds ratios for AD were plotted in the case of increasing Aβ40 percentile classes, and a significant increase ranging from 0.4 to 1.8 was observed (Figure 2B). To establish whether the difference in age observed between NAD and AD patients (SupTable 1) might be a significant determinant here, the age distribution between percentile classes was also plotted (Figure 2B). A significant difference in age distribution was observed only between the 50th/75th and the > 75th Aβ40 percentile classes (Figure 2C). Age cannot therefore account for the association between Aβ40 levels and AD prevalence. Nevertheless further statistical test were adjusted using age as covariate. ### Correlations between Aβ40 and the other CSF biomarkers The correlation between Aβ40 and the other CSF biomarkers was computed in global, NAD and AD populations, for each cohort, and in the overall population (Table 2). As was to be expected [19], a correlation was found to exist between Aβ40 and Aβ42, especially in the NAD group. Aβ40 was also correlated with the tau levels, and it was striking that the highest correlation coefficients were obtained with p-tau (181) rather than with t-tau, especially in the Mtp-1 cohort (a significant difference was observed between the correlation coefficients at P = 0.02). The correlation was clearly visible when the mean-centred p-tau(181) values were plotted in the various Aβ40 percentile classes (Figure 2D), showing significant differences between classes. This correlation had to be put in perspective with the fact that both analytes increased in AD with the patients’age (SupTable 1), which justifies the adjustments made for age in our statistical analysis. We illustrated graphically the correlations in the NAD and the AD populations which had much higher and more widely distributed p-tau(181) values (Figure 3AB). Interestingly, the correlation coefficients were maintained and became even higher in the NAD population. To document the relationship between Aβ40 and p-tau(181) outside the context of AD, these correlation coefficients were tested in a series of clinically defined patients with multiple sclerosis [51] and FTD [52] (Figure 3CD). The corresponding correlation coefficients were both significantly higher in these groups than in the AD population (P< 0.001). ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/06/02/2020.06.02.20119578/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2020/06/02/2020.06.02.20119578/F3) Figure 3: Correlation between Aβ40 and p-tau(181) in different clinical populations To illustrate the correlation between Aβ40 and p-tau(181) (table 2), the mean-centered concentrations of the two analytes in the total study population were plotted in NAD (panel A) and AD populations (panel B). Aβ40 and p-tau(181) concentration were also plotted in a selection of multiple sclerosis (panel C) and FTD patients (panel D). View this table: [Table 2](http://medrxiv.org/content/early/2020/06/02/2020.06.02.20119578/T2) Table 2 Age adjusted Pearson correlation between Aβ40 and Aβ42, tau or p-tau(181) values in the six cohorts (Montpellier 1 (Mtp-1), Montpellier 2 (Mtp-2), Paris, SPIN-Barcelona, ADNI-MS, ADNI-Elecsys), in the overall population using mean-centred values to account for level differences between analytical methods. Computation has been done in the All population and in the NAD, AD, ADNI(-), ADNI(+) groups. Correlation coefficient statistical value P< 0.001 for all but * P< 0.01. ## Discussion The decrease in Aβ42 observed in the CSF of AD patients has attracted considerable attention in clinical and research communities. This decrease is attributable to the accumulation of Aβ42 in the brain parenchyma, along with a decrease in the rates of CSF clearance and an increase in the production of oligomeric/multimeric forms. Since determining the CSF Aβ42/Aβ40 ratio provides a useful means of improving AD diagnosis, many groups are now also measuring Aβ40 in their patients. A meta-analysis was however not conclusive regarding its differential levels in AD [35]. Looking back in detail at various studies, Aβ40 was either lower or showed no significant changes [21, 29, 30, 53], or apparently increased in AD in comparison with other forms of dementia [54, 55]. In a recent report, the increase in Aβ40 was clearly identified as one of the reasons for the good performances of the Aβ42/Aβ40 ratio as an index [20], while another study on PET amyloid findings also established that CSF Aβ40 increased in the PIB+ population [56]. These discrepancies might be linked to differences in cohort composition, since the Aβ40 levels may be affected by various pathological conditions [29–31]. The stage of AD, corresponding to various levels of cerebral atrophy probably reducing amyloid production [57], may also account for differences between studies. This is coherent with a recent study confirming the increase of Aβ40 in prodromal AD [58]. Differences in the precision of the analytical methods used, combined with the size of the cohorts, might also explain why only a small, non-significant difference between AD and NAD patients has been observed in some cases. The present study on Aβ40, which included the largest number of samples studied so far to our knowledge, gave us a sufficiently statistical power to identify small differences. We therefore confirmed the occurrence of an age- and ApoE-independent increase in CSF Aβ40 in AD in comparison with control cohorts consisting mostly of controls and patients with other neurodegenerative diseases and dementia. This observation is valid in different analytical contexts despite differences in the threshold or range for biomarker measurement. It is worth mentioning that in the blood, where the amyloid peptide 42/40 ratio could well indicate the presence of brain amyloidosis [59, 60], it has been established that high Aβ40 levels are associated with greater mortality rate in the elderly [61]. The fact that the CSF and blood amyloid levels are poorly correlated, however, makes it difficult at this stage to extend the present conclusions to this fluid. The overlap in the CSF Aβ40 values between the AD and NAD populations is worth noting, and the area under the receiver operating characteristic curve (AUC) for AD diagnosis was under 0.8 in all the cohorts tested (SupFigure2). CSF Aβ40 cannot therefore be used as a diagnostic biomarker but could be taken to be a feature “risk factor” in view of the odds ratio of almost 2 recorded on the population having the highest CSF Aβ40 concentration. The increased Aβ40 might be a consequences of a reduced clearance of amyloid peptides in sporadic cases and/or a higher production-lower degradation. This matches the fact that in autosomal dominant forms of AD linked to APP or presenilin mutations [3, 5] and in Down syndrome [4], an overproduction of amyloid peptides is thought to trigger the AD process, along with all its consequences, including tau protein hyperphosphorylation, in particular. In this context, baseline Aβ40 concentration could indicate subjects with risk of early AD development. The positive correlation found to exist in the present study between Aβ40 and ptau(181) in AD is an additional argument supporting this pathophysiological model. Tau has many phosphorylated isoforms [62, 63], some of them believed to be more specific for AD than p-tau(181), highlighting the pathophysiological role and therapeutic interest of kinases like PKA, CAMkII or Cdk5. This isoform is however one of the best indicator of AD pathology in the CSF where it begins to increase as two decades before the development of aggregated tau pathology [45]. In this work, we had to rely only on the correlation with p-tau(181) because it is the only isoform with in vitro diagnostic (IVD) certification and has been measured in large clinical cohorts. The fact that this correlation was also present in a control population including a subgroup of well-defined FTD [52] and multiple sclerosis patients [51] raises many questions, however. It is tempting to take this relationship to confirm that Aβ peptides may induce the phosphorylation of tau, as observed both *in vitro* and *in vivo* [64]. Since the present study was based on a cross-sectional design, and without neuropathological confirmation, further studies involving a longitudinal design are now required to confirm the idea that high baseline CSF levels of Aβ peptides may have prejudicial effects, leading to AD. The results obtained here are certainly consistent with the idea that approaches making it possible to reduce amyloid beta production rates and levels in the CSF in particular Aβ40, will create valuable new opportunities for developing new curative and/or preventive interventions in AD. ## Data Availability The datasets used for the analyses are available from the corresponding author on reasonable request. ## Declarations ### Ethics approval and consent to participate *All the patients at each clinical centre gave their written informed consent to participating in clinical research on CSF biomarkers, which was approved by the respective Ethics Committees. The committee responsible in Montpellier was the regional Ethics Committee of the Montpellier University Hospital and Montpellier CSF-Neurobank #DC-2008-417 at the certified NFS 96-900 CHU resource center BB-0033-00031, [www.biobanques.eu](http://www.biobanques.eu). Authorization to handle personal data was granted by the French Data Protection Authority (CNIL) under the number 1709743 v0*. ### Consent for publication *Not applicable*. ### Availability of data and material *The datasets used for the analyses are available from the corresponding author on reasonable request*. ### Competing interests *The authors report no conflict of interest to disclose*. ### Funding *This work was supported by “France Alzheimer” and via the French National Alzheimer effort (“Le Plan Alzheimer”). Data collection and sharing for this project was also funded by the Alzheimer’s Disease Neuroimaging Initiative (ADNI) (National Institutes of Health Grant U01 AG024904) and DOD ADNI (Department of Defense award number W81XWH-12-2-0012). ADNI is funded by the National Institute on Aging, the National Institute of Biomedical Imaging and Bioengineering, and through generous contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica, Inc.; Biogen; Bristol-Myers Squibb Company; CereSpir, Inc.; Cogstate; Eisai Inc.; Elan Pharmaceuticals, Inc.; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche Ltd and its affiliated company Genentech, Inc.; Fujirebio; GE Healthcare; IXICO Ltd.;Janssen Alzheimer Immunotherapy Research & Development, LLC.; Johnson & Johnson Pharmaceutical Research & Development LLC.; Lumosity; Lundbeck; Merck & Co., Inc.;Meso Scale Diagnostics, LLC.; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer Inc.; Piramal Imaging; Servier; Takeda Pharmaceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health ([www.fnih.org](http://www.fnih.org)). The grantee organization is the Northern California Institute for Research and Education, and the study is coordinated by the Alzheimer’s Therapeutic Research Institute at the University of Southern California. ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of Southern California*. ### Authors’ contributions Lehmann, Dumurgier, Lleo, Gabelle had full access to all the data in the study and take responsibility for their presentation and the accuracy of the analysis. Concept and design: Dumurgier, Gabelle, Lehmann. Acquisition of the data (biological and clinical): Ayrignac, Marelli, Alcolea, Fortea, Thouvenot, Delaby, Hirtz, Vialaret, Ginestet, Bouaziz-Amar, Laplanche, Labaug, Paquet. Drafting of the manuscript: Dumurgier, Gabelle, Lehmann. Administrative, technical, and material support: Hirtz, Delaby. ### Abbreviations AD : Alzheimer’s disease ADNI : Alzheimer’s Disease Neuroimaging Initiative APP : amyloid precursor protein AUC : Area under the curve; Aβ: Amyloid β CAA : cerebral amyloid angiopathy CI : Confidence interval CLIA : chemiluminescence immunoassay CMRR : Centre Mémoire de Ressources et de Recherche CSF : Cerebrospinal fluid FTD : Frontotemporal degeneration FTLD : Frontotemporal lobar degeneration LBD : Lewy Body dementia MCI : Mild cognitive impairment MMSE : Mini Mental State Examination MRI : magnetic-resonance brain imaging NAD : Non Alzheimer disease NFT : neurofibrillary tangles PET : Positron-Emission Tomography; PiB: Pittsburgh compound B PLM : Paris-Lille-Montpellier PSP : Progressive Supra-nuclear Palsy QC : Quality control ROC : Receiver operating characteristic SD : Standard deviation. * Received June 2, 2020. * Revision received June 2, 2020. * Accepted June 2, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## Reference 1. 1.Blennow K, de Leon MJ, Zetterberg H: Alzheimer’s disease. The Lancet 2006, 368:387–403. 2. 2.Bloom GS: Amyloid-beta and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol 2014, 71:505–508. 3. 3.Bertram L, Lill CM, Tanzi RE: The genetics of Alzheimer disease: back to the future. Neuron 2010, 68:270–281. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuron.2010.10.013&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20955934&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000284304300015&link_type=ISI) 4. 4.Wiseman FK, Al-Janabi T, Hardy J, Karmiloff-Smith A, Nizetic D, Tybulewicz VL, et al: A genetic cause of Alzheimer disease: mechanistic insights from Down syndrome. Nat Rev Neurosci 2015 16:564–574. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrn3983&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26243569&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 5. 5.Rovelet-Lecrux A, Hannequin D, Raux G, Le Meur N, Laquerriere A, Vital A, et al: APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat Genet 2006, 38:24–26. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng1718&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16369530&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000234227200013&link_type=ISI) 6. 6.Strittmatter WJ, Weisgraber KH, Huang DY, Dong LM, Salvesen GS, Pericak-Vance M, et al: Binding of human apolipoprotein E to synthetic amyloid beta peptide: isoformspecific effects and implications for late-onset Alzheimer disease. Proc Natl Acad Sci U S A 1993, 90:8098–8102. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiOTAvMTcvODA5OCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzA2LzAyLzIwMjAuMDYuMDIuMjAxMTk1NzguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 7. 7.Brothers HM, Gosztyla ML, Robinson SR: The Physiological Roles of Amyloid-beta Peptide Hint at New Ways to Treat Alzheimer’s Disease. Front Aging Neurosci 2018, 10:118. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fnagi.2018.00118&link_type=DOI) 8. 8.Pernegre C, Duquette A, Leclerc N: Tau Secretion: Good and Bad for Neurons. Front Neurosci 2019, 13:649. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fnins.2019.00649&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31293374&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 9. 9.Andreasen N, Blennow K: CSF biomarkers for mild cognitive impairment and early Alzheimer’s disease. Clin Neurol Neurosurg 2005, 107:165–173. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.clineuro.2004.10.011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15823670&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000228678600001&link_type=ISI) 10. 10.Blennow K, Hampel H: CSF markers for incipient Alzheimer’s disease. Lancet Neurol 2003, 2:605–613. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1474-4422(03)00530-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14505582&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000185432000017&link_type=ISI) 11. 11.McKhann GM, Knopman DS, Chertkow H, Hyman BT, Jack CR. Jr..,, Kawas CH, et al: The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011, 7:263–269. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2011.03.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21514250&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000291239600003&link_type=ISI) 12. 12.Dubois B, Feldman HH, Jacova C, Hampel H, Molinuevo JL, Blennow K, et al: Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol 2014, 13:614–629. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1474-4422(14)70090-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24849862&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000337212700024&link_type=ISI) 13. 13.Gabelle A, Dumurgier J, Vercruysse O, Paquet C, Bombois S, Laplanche JL, et al: Impact of the 2008-2012 French Alzheimer Plan on the use of cerebrospinal fluid biomarkers in research memory center: the PLM Study. J Alzheimers Dis 2013, 34:297–305. 14. 14.Blennow K, Dubois B, Fagan AM, Lewczuk P, de Leon MJ, Hampel H: Clinical utility of cerebrospinal fluid biomarkers in the diagnosis of early Alzheimer’s disease. Alzheimers Dement 2014. 15. 15.Lewczuk P, Kornhuber J, Toledo JB, Trojanowski JQ, Knapik-Czajka M, Peters O, et al: Validation of the Erlangen Score Algorithm for the Prediction of the Development of Dementia due to Alzheimer’s Disease in Pre-Dementia Subjects. Journal of Alzheimer’s Disease 2015, 48:433–441. 16. 16.Nordberg A: PET imaging of amyloid in Alzheimer’s disease. Lancet Neurol 2004, 3:519–527. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1474-4422(04)00853-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15324720&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000223568300016&link_type=ISI) 17. 17.Villemagne VL, Fodero-Tavoletti MT, Masters CL,Rowe CC: Tau imaging: early progress and future directions. Lancet Neurol 2015, 14:114–124. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1474-4422(14)70252-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25496902&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 18. 18.Jack CR Jr..,, Wiste HJ, Vemuri P, Weigand SD, Senjem ML, Zeng G, et al: Brain betaamyloid measures and magnetic resonance imaging atrophy both predict time-toprogression from mild cognitive impairment to Alzheimer’s disease. Brain 2010, 133:3336–3348. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/brain/awq277&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20935035&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000283666500022&link_type=ISI) 19. 19.Gabelle A, Roche S, Geny C, Bennys K, Labauge P, Tholance Y, et al: Correlations between soluble alpha/beta forms of amyloid precursor protein and Abeta38, 40, and 42 in human cerebrospinal fluid. Brain Res 2010, 1357:175–183. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.brainres.2010.08.022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20713025&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000283039500020&link_type=ISI) 20. 20.Dorey A, Perret-Liaudet A, Tholance Y, Fourier A, Quadrio I: Cerebrospinal Fluid Abeta40 Improves the Interpretation of Abeta42 Concentration for Diagnosing Alzheimer’s Disease. Front Neurol 2015, 6:247. 21. 21.Lewczuk P, Esselmann H, Otto M, Maler JM, Henkel AW, Henkel MK, et al: Neurochemical diagnosis of Alzheimer’s dementia by CSF Aβ42, Aβ42/Aβ40 ratio and total tau. Neurobiology of Aging 2004, 25:273–281. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0197-4580(03)00086-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15123331&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000189229600001&link_type=ISI) 22. 22.Lehmann S, Delaby C, Boursier G, Catteau C, Ginestet N, Tiers L, et al: Relevance of Abeta42/40 Ratio for Detection of Alzheimer Disease Pathology in Clinical Routine: The PLMR Scale. Front Aging Neurosci 2018, 10:138. 23. 23.Gervaise-Henry C, Watfa G, Albuisson E, Kolodziej A, Dousset B, Olivier JL, et al: Cerebrospinal Fluid Abeta42/Abeta40 as a Means to Limiting Tube-and Storage-Dependent Pre-Analytical Variability in Clinical Setting. J Alzheimers Dis 2017, 57:437–445. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JAD-160865&link_type=DOI) 24. 24.Lewczuk P, Lelental N, Spitzer P, Maler JM, Kornhuber J: Amyloid-beta 42/40 cerebrospinal fluid concentration ratio in the diagnostics of Alzheimer’s disease: validation of two novel assays. J Alzheimers Dis 2015, 43:183–191. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JAD-140771&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25079805&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 25. 25.Willemse E, van Uffelen K, Brix B, Engelborghs S, Vanderstichele H, Teunissen C: How to handle adsorption of cerebrospinal fluid amyloid beta (1–42) in laboratory practice? Identifying problematic handlings and resolving the issue by use of the Abeta42/Abeta40 ratio. Alzheimers Dement 2017, 13:885-892. 26. 26.Hansson O, Lehmann S, Otto M, Zetterberg H, Lewczuk P: Advantages and disadvantages of the use of the CSF Amyloid beta (Abeta) 42/40 ratio in the diagnosis of Alzheimer’s Disease. Alzheimers Res Ther 2019, 11:34. 27. 27.Dumurgier J, Schraen S, Gabelle A, Vercruysse O, Bombois S, Laplanche JL, et al: Cerebrospinal fluid amyloid-beta 42/40 ratio in clinical setting of memory centers: a multicentric study. Alzheimers Res Ther 2015, 7:30. 28. 28.Wiltfang J, Esselmann H, Bibl M, Hull M, Hampel H, Kessler H, et al: Amyloid beta peptide ratio 42/40 but not A beta 42 correlates with phospho-Tau in patients with low-and high-CSF A beta 40 load. J Neurochem 2007, 101:1053-1059. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1471-4159.2006.04404.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17254013&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 29. 29.Bibl M, Mollenhauer B, Lewczuk P, Esselmann H, Wolf S, Otto M, et al: Cerebrospinal fluid tau, p-tau 181 and amyloid-beta38/40/42 in frontotemporal dementias and primary progressive aphasias. Dement Geriatr Cogn Disord 2011, 31:37-44. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1159/000322370&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21135556&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 30. 30.Gabelle A, Roche S, Geny C, Bennys K, Labauge P, Tholance Y, et al: Decreased sAbetaPPbeta, Abeta38, and Abeta40 cerebrospinal fluid levels in frontotemporal dementia. J Alzheimers Dis 2011, 26:553-563. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21709372&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000295032800014&link_type=ISI) 31. 31.Renard D, Castelnovo G, Wacongne A, Le Floch A, Thouvenot E, Mas J, et al: Interest of CSF biomarker analysis in possible cerebral amyloid angiopathy cases defined by the modified Boston criteria. J Neurol 2012. 32. 32.Gisslen M, Krut J, Andreasson U, Blennow K, Cinque P, Brew BJ, et al: Amyloid and tau cerebrospinal fluid biomarkers in HIV infection. BMC Neurol 2009, 9:63. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2377-9-63&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20028512&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 33. 33.Pietroboni AM, Caprioli M, Carandini T, Scarioni M, Ghezzi L, Arighi A, et al: CSF beta-amyloid predicts prognosis in patients with multiple sclerosis. Mult Scler 2019, 25:1223-1231. 34. 34.Chen Z, Liu C, Zhang J, Relkin N, Xing Y, Li Y: Cerebrospinal fluid Abeta42, t-tau, and p-tau levels in the differential diagnosis of idiopathic normal-pressure hydrocephalus: a systematic review and meta-analysis. Fluids Barriers CNS 2017, 14:13. 35. 35.Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, et al: CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. The Lancet Neurology 2016, 15:673–684. 36. 36.Alcolea D, Martinez-Lage P, Sanchez-Juan P, Olazaran J, Antunez C, Izagirre A, et al: Amyloid precursor protein metabolism and inflammation markers in preclinical Alzheimer disease. Neurology 2015 85:626–633. 37. 37.Sala I, Illan-Gala I, Alcolea D, Sanchez-Saudinos MB, Salgado SA, Morenas-Rodriguez E, et al: Diagnostic and Prognostic Value of the Combination of Two Measures of Verbal Memory in Mild Cognitive Impairment due to Alzheimer’s Disease. J Alzheimers Dis 2017, 58:909–918. 38. 38.McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan EM: Clinical diagnosis of Alzheimer’s disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer’s Disease. Neurology 1984, 34:939–944. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.34.7.939&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=6610841&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 39. 39.McKhann GM, Albert MS, Grossman M, Miller B, Dickson D, Trojanowski JQ: Clinical and pathological diagnosis of frontotemporal dementia: report of the Work Group on Frontotemporal Dementia and Pick’s Disease. Arch Neurol 2001, 58:1803–1809. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archneur.58.11.1803&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11708987&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000172055300012&link_type=ISI) 40. 40.McKeith IG: Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the Consortium on DLB International Workshop. J Alzheimers Dis 2006, 9:417–423. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JAD-2006-9S347&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16914880&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 41. 41.Boeve BF, Maraganore DM, Parisi JE, Ahlskog JE, Graff-Radford N, Caselli RJ, et al: Pathologic heterogeneity in clinically diagnosed corticobasal degeneration.Neurology 1999, 53:795–800. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1212/WNL.53.4.795&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10489043&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 42. 42.Lehmann S, Schraen S, Paquet C, Bombois S, Delaby C, Dorey A, et al: A diagnostic scale for Alzheimer’s disease based on cerebrospinal fluid biomarker profiles. Alzheimer’s Research & Therapy 2014, 6:38 43. 43.Dumurgier J, Vercruysse O, Paquet C, Bombois S, Chaulet C, Laplanche JL, et al: Intersite variability of CSF Alzheimer’s disease biomarkers in clinical setting. Alzheimers Dement 2013, 9:406–413. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jalz.2012.06.006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23141384&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000321729000007&link_type=ISI) 44. 44.Del Campo M, Mollenhauer B, Bertolotto A, Engelborghs S, Hampel H, Simonsen AH, et al: Recommendations to standardize preanalytical confounding factors in Alzheimer’s and Parkinson’s disease cerebrospinal fluid biomarkers: an update. Biomark Med 2012, 6:419–430. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2217/bmm.12.46&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22917144&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 45. 45.Barthelemy NR, Li Y, Joseph-Mathurin N, Gordon BA, Hassenstab J, Benzinger TLS, et al: A soluble phosphorylated tau signature links tau, amyloid and the evolution of stages of dominantly inherited Alzheimer’s disease. Nat Med 2020 26:398–407. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-0781-z&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32161412&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 46. 46.Hansson O, Seibyl J, Stomrud E, Zetterberg H, Trojanowski JQ, Bittner T, et al: CSF biomarkers of Alzheimer’s disease concord with amyloid-beta PET and predict clinical progression: A study of fully automated immunoassays in BioFINDER and ADNI cohorts. Alzheimers Dement 2018, 14:1470–1481. 47. 47.Spellman DS, Wildsmith KR, Honigberg LA, Tuefferd M, Baker D, Raghavan N, et al: Development and evaluation of a multiplexed mass spectrometry based assay for measuring candidate peptide biomarkers in Alzheimer’s Disease Neuroimaging Initiative (ADNI) CSF. Proteomics Clin Appl 2015, 9:715–731. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/prca.201400178&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25676562&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 48. 48.Perret-Liaudet A, Pelpel M, Tholance Y, Dumont B, Vanderstichele H, Zorzi W, et al: Risk of Alzheimer’s disease biological misdiagnosis linked to cerebrospinal collection tubes. J Alzheimers Dis 2012, 31:13–20. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JAD-2012-120361&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22495345&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000306122900003&link_type=ISI) 49. 49.Peskind ER, Li G, Shofer J, Quinn JF, Kaye JA, Clark CM, et al: Age and apolipoprotein E*4 allele effects on cerebrospinal fluid beta-amyloid 42 in adults with normal cognition. Arch Neurol 2006, 63:936–939. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archneur.63.7.936&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16831961&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 50. 50.Sunderland T, Mirza N, Putnam KT, Linker G, Bhupali D, Durham R, et al: Cerebrospinal fluid beta-amyloid1-42 and tau in control subjects at risk for Alzheimer’s disease: the effect of APOE epsilon4 allele. Biol Psychiatry 2004, 56:670–676. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2004.07.021&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15522251&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000224994900009&link_type=ISI) 51. 51.Vukusic S, Casey R, Rollot F, Brochet B, Pelletier J, Laplaud DA, et al: Observatoire Francais de la Sclerose en Plaques (OFSEP): A unique multimodal nationwide MS registry in France. Mult Scler 2018:1352458518815602. 52. 52.Marelli C, Gutierrez LA, Menjot de Champfleur N, Charroud C,De Verbizier D, Touchon J, et al: Late-onset behavioral variant of frontotemporal lobar degeneration versus Alzheimer’s disease: Interest of cerebrospinal fluid biomarker ratios. Alzheimers Dement (Amst) 2015, 1:371–379. 53. 53.Janelidze S, Zetterberg H, Mattsson N, Palmqvist S, Vanderstichele H, Lindberg O, et al: CSF Abeta42/Abeta40 and Abeta42/Abeta38 ratios: better diagnostic markers of Alzheimer disease. Ann Clin Transl Neurol 2016, 3:154–165. 54. 54.Welge V, Fiege O, Lewczuk P, Mollenhauer B, Esselmann H, Klafki HW, et al: Combined CSF tau, p-tau181 and amyloid-beta 38/40/42 for diagnosing Alzheimer’s disease. J Neural Transm 2009, 116:203–212. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00702-008-0177-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19142572&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000269823900011&link_type=ISI) 55. 55.Sundelof J, Sundstrom J, Hansson O, Eriksdotter-Jonhagen M, Giedraitis V, Larsson A, et al: Higher cathepsin B levels in plasma in Alzheimer’s disease compared to healthy controls. J Alzheimers Dis 2010, 22:1223–1230. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20930303&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 56. 56.Lewczuk P, Matzen A, Blennow K, Parnetti L, Molinuevo JL, Eusebi P, et al: Cerebrospinal Fluid Abeta42/40 Corresponds Better than Abeta42 to Amyloid PET in Alzheimer’s Disease. J Alzheimers Dis 2017, 55:813–822. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3233/JAD-160722&link_type=DOI) 57. 57.Jack CR Jr..,, Wiste HJ, Vemuri P, Weigand SD, Senjem ML, Zeng G, et al: Brain betaamyloid measures and magnetic resonance imaging atrophy both predict time-toprogression from mild cognitive impairment to Alzheimer’s disease. Brain 2011, 133:3336–3348. 58. 58.Tijms BM, Vermunt L, Zwan MD, van Harten AC, van der Flier WM, Teunissen CE, et al: Pre-amyloid stage of Alzheimer’s disease in cognitively normal individuals. Ann Clin Transl Neurol 2018, 5:1037–1047. 59. 59.Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Dore V, et al: High performance plasma amyloid-beta biomarkers for Alzheimer’s disease. Nature 2018, 554:249–254. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature25456&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29420472&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 60. 60.Hanon O, Vidal JS, Lehmann S, Bombois S, Allinquant B, Treluyer JM, et al: Plasma amyloid levels within the Alzheimer’s process and correlations with central biomarkers. Alzheimers Dement 2018. 61. 61.Gabelle A, Schraen S, Gutierrez LA, Pays C, Rouaud O, Buee L, et al: Plasma betaamyloid 40 levels are positively associated with mortality risks in the elderly. Alzheimers Dement 2015, 11:672–680. 62. 62.Simic G, Babic Leko M, Wray S, Harrington C, Delalle I,Jovanov-Milosevic N, et al: Tau Protein Hyperphosphorylation and Aggregation in Alzheimer’s Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules 2016 6:6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/biom6010006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26751493&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 63. 63.Martin L, Latypova X, Terro F: Post-translational modifications of tau protein: implications for Alzheimer’s disease. Neurochem Int 2011, 58:458–471. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuint.2010.12.023&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21215781&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F06%2F02%2F2020.06.02.20119578.atom) 64. 64.Selenica ML, Brownlow M, Jimenez JP, Lee DC, Pena G, Dickey CA, et al: Amyloid oligomers exacerbate tau pathology in a mouse model of tauopathy. Neurodegener Dis 2013, 11:165–181.